Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Junghee Kang is active.

Publication


Featured researches published by Junghee Kang.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Akt2 overexpression plays a critical role in the establishment of colorectal cancer metastasis

Piotr G. Rychahou; Junghee Kang; Pat Gulhati; Hung Q. Doan; L. Andy Chen; Shu-Yuan Xiao; Dai H. Chung; B. Mark Evers

Colorectal cancer is the second leading cause of cancer-related deaths in the United States. Understanding the distinct genetic and epigenetic changes contributing to the establishment and growth of metastatic lesions is crucial for the development of novel therapeutic strategies. In a search for key regulators of colorectal cancer metastasis establishment, we have found that the serine/threonine kinase Akt2, a known proto-oncogene, is highly expressed in late-stage colorectal cancer and metastatic tumors. Suppression of Akt2 expression in highly metastatic colorectal carcinoma cells inhibits their ability to metastasize in an experimental liver metastasis model. Overexpression of wild-type Akt1 did not restore metastatic potential in cells with downregulated Akt2, thus suggesting non-redundant roles for the individual Akt isoforms. In contrast, Akt2 overexpression in wild-type PTEN expressing SW480 colorectal cancer cells led to the formation of micrometastases; however, loss of PTEN is required for sustained formation of overt metastasis. Finally, we found that the consequence of PTEN loss and Akt2 overexpression function synergistically to promote metastasis. These results support a role for Akt2 overexpression in metastatic colorectal cancer and establish a mechanistic link between Akt2 overexpression and PTEN mutation in metastatic tumor establishment and growth. Taken together, these data suggest that Akt family members have distinct functional roles in tumor progression and that selective targeting of the PI3K/Akt2 pathway may provide a novel treatment strategy for colorectal cancer metastasis.


International Journal of Cancer | 2003

Geldanamycin decreases Raf-1 and Akt levels and induces apoptosis in neuroblastomas

Sunghoon Kim; Junghee Kang; Wanqin Hu; B. Mark Evers; Dai H. Chung

Neuroblastomas are the most common extracranial solid tumors of childhood. These tumors are associated with an overall poor prognosis, particularly for advanced stage disease. The benzoquinone ansamycin antibiotic, geldanamycin (GA), exhibits potent antitumor activity in certain cancer cell lines by destabilizing important signal transduction proteins (e.g., Raf‐1 and Akt). The purpose of our study was to determine whether GA can alter the expression of Raf‐1 and Akt, which have been shown to be critical for neuronal cell survival, and induce apoptosis of neuroblastoma cells. Human neuroblastoma cells (SH‐SY5Y, SK‐N‐SH and LAN‐1) were treated with GA for a variable period of time. Cell viability was assessed with MTT assays. Apoptosis was assessed with DNA fragmentation ELISA, TUNEL‐flow cytometric assay, Western blot and caspase activities. We found that GA decreases cell viability and induces apoptosis in the SH‐SY5Y human neuroblastoma cell line. These effects were mediated through activation of caspase‐9 and ‐3, mitochondrial release of cytochrome c and subsequent PARP cleavage. GA‐induced apoptosis was associated with a reduction in the level and activity of Raf‐1 and Akt. The importance of these proteins was further demonstrated by induction of apoptosis in SH‐SY5Y cells by a combination of U0126 (MEK1/2 inhibitor) and LY294002 (an inhibitor of PI3K). Similar to SH‐SY5Y cells, other human neuroblastoma cells (SK‐N‐SH and LAN‐1) were sensitive to the effects of GA‐induced apoptosis. Taken together, our findings suggest that GA may be a novel therapeutic agent, which may be effective in the treatment of neuroblastomas.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Gastrin-releasing peptide receptor silencing suppresses the tumorigenesis and metastatic potential of neuroblastoma

Jingbo Qiao; Junghee Kang; Titilope A. Ishola; Piotr G. Rychahou; B. Mark Evers; Dai H. Chung

Neuroblastoma accounts for nearly 15% of all pediatric cancer-related deaths. We have previously shown that gastrin-releasing peptide (GRP) stimulates neuroblastoma growth, and that its cell surface receptor, GRP-R, is overexpressed in advanced-stage human neuroblastomas; however, the effects of GRP/GRP-R on tumorigenesis and metastasis in vivo are not clearly elucidated. In the present study, we found that GRP-R knockdown in the aggressive cell line BE(2)-C induced cell morphology changes, reduced cell size, decreased cell proliferation, and inhibited DNA synthesis, corresponding to cell cycle arrest at G2/M phase. Activated Akt, a crucial regulator of cell survival and metastasis, was down-regulated by GRP-R silencing. In addition, expression of p-p70S6K and its downstream target molecule S6, key regulators of protein synthesis and cell metabolism, were also significantly decreased by GRP-R silencing. GRP-R knockdown also up-regulated the expression of tumor suppressor PTEN, the inhibitor of the PI3K/Akt pathway. Furthermore, silencing GRP-R as well as GRP in BE(2)-C cells suppressed anchorage-independent growth in vitro. Conversely, overexpression of GRP-R in less aggressive SK-N-SH neuroblastoma cells resulted in soft agar colony formation, which was inhibited by a GRP-blocking antibody. Moreover, GRP-R deficiency significantly delayed tumor growth and diminished liver metastases in vivo. Our findings demonstrate that GRP and GRP-R have important oncogenic properties beyond their established mitogenic functions. Therefore, GRP-R may be an ideal therapeutic target for the treatment of aggressive neuroblastomas.


Annals of Surgery | 2005

Gastrin-releasing peptide-induced down-regulation of tumor suppressor protein PTEN (phosphatase and tensin homolog deleted on chromosome ten) in neuroblastomas.

Jingbo Qiao; Junghee Kang; Jeremy Cree; B. Mark Evers; Dai H. Chung

Objectives:To evaluate whether aggressive, undifferentiated neuroblastomas express tumor suppressor protein PTEN (phosphatase and tensin homolog deleted on chromosome ten) and to examine the effects of gastrin-releasing peptide (GRP) on PTEN gene and protein expression. Summary Background Data:We have previously shown that neuroblastomas secrete GRP, which binds to its cell surface receptor (GRP-R) to stimulate cell growth in an autocrine fashion. However, the effects of GRP on expression of the tumor suppressor gene PTEN have not been elucidated in neuroblastomas. Methods:Paraffin-embedded sections from human neuroblastomas were analyzed for PTEN and phospho-Akt protein expression by immunohistochemistry. Human neuroblastoma cell lines (SK-N-SH and SH-SY5Y) were stably transfected with the plasmid pEGFP-GRP-R to establish GRP-R overexpression cell lines, and the effects of GRP on PTEN gene and protein expression were determined. Results:A decrease in the ratio of PTEN to phospho-Akt protein expression was identified in poorly differentiated neuroblastomas. An increase in GRP binding capacity was confirmed in GRP-R overexpressing cells, which demonstrated an accelerated constitutive cell growth rate. PTEN gene and protein expression was significantly decreased in GRP-R overexpressing cells when compared with controls. Conclusions:Our findings demonstrate decreased expression of the tumor suppressor protein PTEN in more aggressive undifferentiated neuroblastomas. An increase in GRP binding capacity, as a result of GRP-R overexpression, down-regulates PTEN expression. These findings suggest that an inhibition of the tumor suppressor gene PTEN may be an important regulatory mechanism involved in GRP-induced cell proliferation in neuroblastomas.


Surgery | 2011

Targeting gastrin-releasing peptide as a new approach to treat aggressive refractory neuroblastomas

Pritha Paul; Lauren A. Gillory; Junghee Kang; Jingbo Qiao; Dai H. Chung

BACKGROUND The overall survival for neuroblastoma remains dismal, in part due to the emergence of resistance to chemotherapeutic drugs. We have demonstrated that gastrin-releasing peptide (GRP), a gut peptide secreted by neuroblastoma, acts as an autocrine growth factor. We hypothesized that knockdown of GRP will induce apoptosis in neuroblastoma cells and potentiate the cytotoxic effects of chemotherapeutic agents. METHODS The human neuroblastoma cell lines (JF, SK-N-SH) were transfected with small interfering (si) RNA targeted at GRP. Apoptosis was assessed by DNA fragmentation assay. Immunoblotting was used to confirm molecular markers of apoptosis, and flow cytometry was performed to determine cell cycle arrest after GRP knockdown. RESULTS siGRP resulted in an increase in apoptosis in the absence of chemotherapeutic interventions. A combination of GRP silencing and chemotherapeutic drugs resulted in enhanced apoptosis when compared to either of the treatments alone. GRP silencing led to increased expression of proapoptotic proteins, p53 and p21. CONCLUSION Silencing of GRP induces apoptosis in neuroblastoma cells; it acts synergistically with chemotherapeutic effects of etoposide and vincristine. GRP knockdown-mediated apoptosis appears to be associated with upregulation of p53 in neuroblastoma cells. Targeting GRP may be postulated as a potential novel agent for combinational treatment to treat aggressive neuroblastomas.


Journal of Biological Chemistry | 2004

Down-regulation of the Tumor Suppressor PTEN by the Tumor Necrosis Factor-α/Nuclear Factor-κB (NF-κB)-inducing Kinase/NF-κB Pathway Is Linked to a Default IκB-α Autoregulatory Loop

Sunghoon Kim; Claire Domon-Dell; Junghee Kang; Dai H. Chung; Jean-Noël Freund; B. Mark Evers


Journal of Biological Chemistry | 2003

Down-regulation of the tumor suppressor PTEN by the TNFα/NIK/NF-κB pathway is linked to a default IκB-α autoregulatory loop

Sunghoon Kim; Claire Domon-Dell; Junghee Kang; Dai H. Chung; Jean-Noël Freund; B. Mark Evers


Biochemical and Biophysical Research Communications | 2006

MYCN silencing induces differentiation and apoptosis in human neuroblastoma cells

Junghee Kang; Piotr G. Rychahou; Titilope A. Ishola; Jingbo Qiao; B. Mark Evers; Dai H. Chung


Journal of Pediatric Surgery | 2004

Phosphatidylinositol 3-kinase inhibition down-regulates survivin and facilitates TRAIL-mediated apoptosis in neuroblastomas

Sunghoon Kim; Junghee Kang; Jingbo Qiao; Robert P. Thomas; B. Mark Evers; Dai H. Chung


Cancer Letters | 2007

Bombesin induces angiogenesis and neuroblastoma growth

Junghee Kang; Titilope A. Ishola; Naira Baregamian; Joshua M. Mourot; Piotr G. Rychahou; B. Mark Evers; Dai H. Chung

Collaboration


Dive into the Junghee Kang's collaboration.

Top Co-Authors

Avatar

Dai H. Chung

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jingbo Qiao

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Titilope A. Ishola

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

B. Evers

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeremy Cree

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Hung Q. Doan

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Joshua M. Mourot

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge